Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 10(1): 4561, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32165710

ABSTRACT

Insulin resistance is associated with aging in mice and humans. We have previously shown that administration of recombinant GDF11 (rGDF11) to aged mice alters aging phenotypes in the brain, skeletal muscle, and heart. While the closely related protein GDF8 has a role in metabolism, limited data are available on the potential metabolic effects of GDF11 or GDF8 in aging. To determine the metabolic effects of these two ligands, we administered rGDF11 or rGDF8 protein to young or aged mice fed a standard chow diet, short-term high-fat diet (HFD), or long-term HFD. Under nearly all of these diet conditions, administration of exogenous rGDF11 reduced body weight by 3-17% and significantly improved glucose tolerance in aged mice fed a chow (~30% vs. saline) or HF (~50% vs. saline) diet and young mice fed a HFD (~30%). On the other hand, exogenous rGDF8 showed signifcantly lesser effect or no effect at all on glucose tolerance compared to rGDF11, consistent with data demonstrating that GFD11 is a more potent signaling ligand than GDF8. Collectively, our results show that administration of exogenous rGDF11, but not rGDF8, can reduce diet-induced weight gain and improve metabolic homeostasis.


Subject(s)
Aging/metabolism , Body Weight/drug effects , Bone Morphogenetic Proteins/administration & dosage , Diet, High-Fat/adverse effects , Insulin Resistance , Myostatin/administration & dosage , Aging/blood , Aging/drug effects , Animals , Bone Morphogenetic Proteins/pharmacology , Energy Metabolism/drug effects , Growth Differentiation Factors/administration & dosage , Growth Differentiation Factors/pharmacology , Male , Mice , Mice, Inbred C57BL , Myostatin/pharmacology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
2.
Theriogenology ; 101: 123-134, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28708509

ABSTRACT

Growth Differentiation Factor 8 (GDF8) is a member of the transforming growth factor-ß (TGF-ß) family and has been identified as a strong physiological regulator. This factor is expressed as a paracrine factor in mural granulosa cells. To investigate the effects of GDF8 on the in vitro maturation (IVM) of porcine oocytes, we assessed the quality of matured oocytes as well as the specific gene transcription and protein activation levels in oocytes and cumulus cells (CCs) after IVM and subsequent embryonic development after in vitro fertilization (IVF) and parthenogenetic activation (PA). Supplemental concentrations (0, 1, 10, and 100 ng/ml) of GDF8 were provided in IVM medium. Supplementation with GDF8 during IVM induced transcription of specific TGF-ß receptor genes, such as ActRIIb and Alk4/5, and the recognition of the GDF8 by these receptors induced phosphorylation of p38 MAPK. Activated p38 MAPK signaling changed oocyte maturation and cumulus expansion-related gene transcription: Nrf2 and Bcl-2 in oocytes and PCNA, Nrf2, Has2, Ptx3, and TNFAIP6 in CCs. The altered gene expression pattern during IVM resulted in a 10% lower level of intracellular ROS in mature oocytes. The improved cytoplasmic maturation led to an increase in the fertilization efficiency and subsequent embryonic developmental competence. The embryonic development showed increases in the blastocyst formation rate and higher transcription levels of POU5F1 and BCL-2 in the blastocysts. The present study suggests that supplementation of GDF8 during IVM synergistically improved the developmental potential of IVF- and PA-derived porcine embryos by reducing the intracellular ROS level in oocytes by altering the transcription of specific genes and increasing the phosphorylation of p38 MAPK during IVM. In conclusion, for the first time, our results demonstrate that GDF8 can act as a paracrine factor to modulate oocyte maturation by regulating p38 MAPK phosphorylation and intracellular ROS level during porcine IVM.


Subject(s)
In Vitro Oocyte Maturation Techniques/veterinary , Myostatin/administration & dosage , Signal Transduction/drug effects , Sus scrofa , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Blastocyst/physiology , Culture Media , Cumulus Cells/physiology , Embryonic Development/drug effects , Female , Fertilization in Vitro/methods , Fertilization in Vitro/veterinary , Gene Expression/drug effects , In Vitro Oocyte Maturation Techniques/methods , Myostatin/genetics , Oocytes/chemistry , Oocytes/drug effects , Oocytes/physiology , Parthenogenesis/drug effects , Phosphorylation/drug effects , Reactive Oxygen Species/analysis , Recombinant Proteins/administration & dosage , p38 Mitogen-Activated Protein Kinases/drug effects
3.
Genet Mol Res ; 15(2)2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27420960

ABSTRACT

Prokaryotic expression technology was used to express maltose-binding protein binding myostatin (MSTN) propeptide fusion protein. Six disease-free Altay lambs were used in this study. The right leg gastrocnemii were injected with MSTN recombinant propeptide protein. The left leg gastrocnemii (the control group) were injected with the same dose of phosphate based saline. The lambs were fed during four months under the same conditions and then slaughtered. Gastrocnemius samples were hematoxylin-eosin stained and the size of the muscle fibers was measured. A real-time polymerase chain reaction (RT-PCR) showed that single gastrocnemius cells in the experimental group had an average area of 1163.01 µm(2), while it was 845.09 µm(2) in the control group (P < 0.05). This indicates that the MSTN propeptide biological agents had an inhibitory effect on MSTN. In order to reveal its mechanism, RT-PCR was conducted to detect the expression of the differentiation-associated genes MyoD, Myf5, Myogenin, p21, and Smad3. The results showed that, in the MSTN propeptide biological agent injected group, expression levels of MSTN, Smad3, and p21 were lower than the control group, while Myf5, MyoD, and Myogenin were higher compared to the control group. This indicates that, when expression of the MSTN gene was inhibited, muscle cell differentiation and growth can be promoted by Smad3 up-regulated expression of Myf5, MyoD, and Myogenin.


Subject(s)
Muscle, Skeletal/drug effects , Myostatin/pharmacology , Sheep/growth & development , Animals , Female , Injections, Intramuscular , Male , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenin/genetics , Myogenin/metabolism , Myostatin/administration & dosage , Sheep/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism
4.
Rejuvenation Res ; 12(2): 85-94, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19405813

ABSTRACT

Myostatin is a member of the transformating growth factor-beta (TGF-beta) superfamily of proteins and is produced almost exclusively in skeletal muscle tissue, where it is secreted and circulates as a serum protein. Myostatin acts as a negative regulator of muscle mass through the canonical SMAD2/3/4 signaling pathway. Naturally occurring myostatin mutants exhibit a 'double muscling' phenotype in which muscle mass is dramatically increased as a result of both hypertrophy and hyperplasia. Myostatin is naturally inhibited by its own propeptide; therefore, we assessed the impact of adeno-associated virus-8 (AAV8) myostatin propeptide vectors when systemically introduced in MF-1 mice. We noted a significant systemic increase in muscle mass in both slow and fast muscle phenotypes, with no evidence of hyperplasia; however, the nuclei-to- cytoplasm ratio in all myofiber types was significantly reduced. An increase in muscle mass in slow (soleus) muscle led to an increase in force output; however, an increase in fast (extensor digitorum longus [EDL]) muscle mass did not increase force output. These results suggest that the use of gene therapeutic regimens of myostatin inhibition for age-related or disease-related muscle loss may have muscle-specific effects.


Subject(s)
Dependovirus/metabolism , Gene Transfer Techniques , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Myostatin/administration & dosage , Peptides/administration & dosage , Protein Precursors/administration & dosage , Animals , Biomechanical Phenomena/drug effects , Body Weight/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Hyperplasia , Hypertrophy , Injections, Intravenous , Mice , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/drug effects , Muscle Fibers, Slow-Twitch/pathology , Myostatin/antagonists & inhibitors , Myostatin/metabolism , Myostatin/pharmacology , Organ Size/drug effects , Peptides/metabolism , Peptides/pharmacology , Protein Precursors/metabolism , Protein Precursors/pharmacology , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/pathology
5.
Hum Gene Ther ; 20(1): 1-10, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18828709

ABSTRACT

Inhibition or blockade of myostatin, a negative growth factor of skeletal muscle, enhances muscle growth and therefore is considered a promising strategy for the treatment of muscle-wasting diseases such as the muscular dystrophies. Previously, we showed that myostatin blockade in both normal and dystrophin-deficient mdx mice by systemic delivery of the myostatin propeptide (MPRO) gene by an adeno-associated virus serotype 8 (AAV8) vector could enhance muscle growth and ameliorate dystrophic lesions. Here, we further investigate whether the muscle growth effect of myostatin blockade can be achieved in dogs by gene transfer. First, we cloned the canine MPRO gene, packaged it in the AAV8 vector, and showed robust muscle-enhancing effects after systemic delivery into neonatal mice. This vector was then further tested in two 3-month-old normal dogs (weighing 9.7 and 6.3 kg). The vector was delivered to one limb by hydrodynamic vein injection, and the contralateral limb served as a control. The delivery procedure was safe, without discernible adverse effects. AAV vector DNA and MPRO gene expression were detected by quantitative polymerase chain reaction, Western blotting, and immunofluorescence staining of muscle biopsies. Overexpression of MPRO resulted in enhanced muscle growth without a cytotoxic T lymphocytic immune response, as evidenced by larger myofibers in multiple muscles, increased muscle volume determined by magnetic resonance imaging, and the lack of CD4+ and CD8+ T cell infiltration in the vector-injected limbs. Our preliminary study thus supports further investigation of this therapeutic strategy in the dystrophin-deficient golden retriever muscular dystrophy dog model.


Subject(s)
Dependovirus/genetics , Genetic Vectors/administration & dosage , Muscle, Skeletal/growth & development , Myostatin/administration & dosage , Peptides/administration & dosage , Animals , Dependovirus/classification , Dependovirus/metabolism , Dogs , Gene Transfer Techniques , Genetic Vectors/genetics , Injections, Intravenous/methods , Male , Mice , Muscle Development/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myostatin/genetics , Myostatin/metabolism , Myostatin/pharmacology , Peptides/genetics , Peptides/metabolism , Peptides/pharmacology , Serotyping
6.
Anim Sci J ; 80(5): 585-90, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20163624

ABSTRACT

The study was conducted to investigate the effects of active immunization against myostatin on the titer of myostatin antibody, carcass evaluation, activity of creatine kinase and the expression of the myostatin gene in pigs. Eighteen pigs were allotted into three groups (six pigs per group), and pigs in treatment 1, 2 and 3 were immunized with physiological saline, 1 mg or 4 mg myostatin per pig, respectively. Six pigs were killed by electrical stunning followed by exsanguination at BW of 100 kg. The results indicated that the titer of myostatin antibody was increased in treated groups compared to the control group on day 42 (P < 0.01) and d 84 (P < 0.01). The carcass lean percentage was significantly increased in the treatment groups compared to the control group (P < 0.01), and intramuscular fat was significantly decreased in the 4 mg group compared to the control group (P < 0.05). The muscle creatine kinase activity of pigs treated with 1 mg and 4 mg myostatin was lower than the control group. The immunization of myostatin significantly decreased the myostatin gene expression levels in muscle. It was concluded that optimal active immunization against myostatin could increase the content of myostatin antibody, suppress the activity of creatine kinase and the expression of myostatin gene, and therefore improve the carcass lean percentage for pigs.


Subject(s)
Antibodies/blood , Creatine Kinase/metabolism , Muscle, Skeletal/metabolism , Myostatin/antagonists & inhibitors , Myostatin/genetics , Myostatin/immunology , Adipose Tissue, White/growth & development , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Animal Husbandry , Animals , Antibodies/immunology , Down-Regulation/immunology , Immunization , Injections, Intramuscular , Meat , Muscle, Skeletal/growth & development , Muscle, Skeletal/immunology , Myostatin/administration & dosage , Sus scrofa
SELECTION OF CITATIONS
SEARCH DETAIL
...